Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cardiovasc Res ; 106(3): 398-407, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25852081

RESUMEN

AIMS: Increased myocardial wall strain triggers the cardiac hypertrophic response by increasing cardiomyocyte size, reprogramming gene expression, and enhancing contractile protein synthesis. The LIM protein, migfilin, is a cytoskeleton-associated protein that was found to translocate in vitro into the nucleus in a Ca(2+)-dependent manner, where it co-activates the pivotal cardiac transcription factor Csx/Nkx2.5. However, the in vivo role of migfilin in cardiac function and stress response is unclear. METHODS AND RESULTS: To define the role of migfilin in cardiac hypertrophy, we induced hypertension by transverse aortic constriction (TAC) and compared cardiac morphology and function of migfilin knockout (KO) with wild-type (WT) hearts. Heart size and myocardial contractility were comparable in untreated migfilin KO and WT hearts, but migfilin-null hearts presented a reduced extent of hypertrophic remodelling in response to chronic hypertensile stress. Migfilin KO mice maintained their cardiac function for a longer time period compared with WT mice, which presented extensive fibrosis and death due to heart failure. Migfilin translocated into the nucleus of TAC-treated cardiomyocytes, and migfilin KO hearts showed reduced Akt activation during the early response to pressure overload. CONCLUSIONS: Our findings indicate an important role of migfilin in the regulation of cardiac hypertrophy upon experimental TAC.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hipertensión/complicaciones , Hipertrofia Ventricular Izquierda/metabolismo , Miocitos Cardíacos/metabolismo , Remodelación Ventricular , Transporte Activo de Núcleo Celular , Animales , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Modelos Animales de Enfermedad , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Hipertensión/fisiopatología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/prevención & control , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factores de Tiempo
2.
Cell Cycle ; 13(3): 462-70, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24280829

RESUMEN

The spatiotemporal manipulations of gene expression by the Cre recombinase (Cre) of bacteriophage P1 has become an essential asset to understanding mammalian genetics. Accumulating evidence suggests that Cre activity can, in addition to excising targeted loxP sites, induce cytotoxic effects, including abnormal cell cycle progression, genomic instability, and apoptosis, which can accelerate cancer progression. It is speculated that these defects are caused by Cre-induced DNA damage at off-target sites. Here we report the formation of tetraploid keratinocytes in the epidermis of keratin 5 and/or keratin 14 promoter-driven Cre (KRT5- and KRT14-Cre) expressing mouse skin. Biochemical analyses and flow cytometry demonstrated that Cre expression also induces DNA damage, genomic instability, and tetraploidy in HCT116 cells, and live-cell imaging revealed an extension of the G 2 cell cycle phase followed by defective or skipping of mitosis as cause for the tetraploidy. Since tetraploidy eventually leads to aneuploidy, a hallmark of cancer, our findings highlight the importance of distinguishing non-specific cytopathic effects from specific Cre/loxP-driven genetic manipulations when using Cre-mediated gene deletions.


Asunto(s)
Daño del ADN , Integrasas/metabolismo , Tetraploidía , Proteínas Virales/metabolismo , Animales , Bacteriófago P1/enzimología , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Citocinesis , Células Epidérmicas , Puntos de Control de la Fase G2 del Ciclo Celular , Inestabilidad Genómica , Células HCT116 , Humanos , Integrasas/genética , Queratina-14/genética , Queratina-5/genética , Queratinocitos/citología , Ratones Transgénicos , Mitosis , Regiones Promotoras Genéticas , Proteínas Virales/genética
3.
J Biol Chem ; 288(26): 18863-71, 2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23658024

RESUMEN

Integrin-linked kinase (ILK) localizes to focal adhesions (FAs) where it regulates cell spreading, migration, and growth factor receptor signaling. Previous reports showed that overexpressed ILK in which Val(386) and Thr(387) were substituted with glycine residues (ILK-VT/GG) could neither interact with paxillin nor localize to FA in cells expressing endogenous wild-type ILK, implying that paxillin binding to ILK is required for its localization to FAs. Here, we show that introducing this mutation into the germ line of mice (ILK-VT/GG) caused vasculogenesis defects, resulting in a general developmental delay and death at around embryonic day 12.5. Fibroblasts isolated from ILK-VT/GG mice contained mutant ILK in FAs, showed normal adhesion to and spreading on extracellular matrix substrates but displayed impaired migration. Biochemical analysis revealed that VT/GG substitutions decreased ILK protein stability leading to decreased ILK levels and reduced binding to paxillin and α-parvin. Because paxillin depletion did not affect ILK localization to FAs, the embryonic lethality and the in vitro migration defects are likely due to the reduced levels of ILK-VT/GG and diminished binding to parvins.


Asunto(s)
Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica , Mutación , Paxillin/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Adhesión Celular , Movimiento Celular , Citometría de Flujo , Adhesiones Focales/metabolismo , Genes Letales , Ratones , Proteínas de Microfilamentos/metabolismo , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Factores de Tiempo
4.
J Cell Sci ; 125(Pt 23): 5690-701, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22992463

RESUMEN

Efficient wound repair is essential for the maintenance of the integrity of the skin. The repair process is controlled by a variety of growth factors and cytokines, and their abnormal expression or activity can cause healing disorders. Here, we show that wound repair is severely delayed in mice lacking fibroblast growth factor receptors (FGFR) 1 and 2 in keratinocytes. As the underlying mechanism, we identified impaired wound contraction and a delay in re-epithelialization that resulted from impaired keratinocyte migration at the wound edge. Scratch wounding and transwell assays demonstrated that FGFR1/2-deficient keratinocytes had a reduced migration velocity and impaired directional persistence owing to inefficient formation and turnover of focal adhesions. Underlying this defect, we identified a significant reduction in the expression of major focal adhesion components in the absence of FGFR signaling, resulting in a general migratory deficiency. These results identify FGFs as key regulators of keratinocyte migration in wounded skin.


Asunto(s)
Queratinocitos/citología , Queratinocitos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Piel/metabolismo , Piel/patología , Cicatrización de Heridas/fisiología , Animales , Western Blotting , Adhesión Celular/genética , Adhesión Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Células Cultivadas , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cicatrización de Heridas/genética
5.
J Cell Sci ; 124(Pt 3): 414-21, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21224394

RESUMEN

Migfilin is a LIM-domain-containing protein of the zyxin family of adaptor proteins and is found at cell-matrix and cell-cell adhesion sites and in the nucleus. In vitro studies have suggested that migfilin promotes ß1 integrin activity, regulates cell spreading and migration and induces cardiomyocyte differentiation. To test directly the function of migfilin in vivo, we generated a migfilin-null mouse strain. Here, we report that loss of migfilin expression permits normal development and normal postnatal aging. Fibroblasts and keratinocytes from migfilin-null mice display normal spreading and adhesion, and normal integrin expression and activation. The migration velocity and directionality of migfilin-null embryonic fibroblasts were normal, whereas the velocity of migfilin-null keratinocytes in wound scratch assays was slightly but significantly reduced. Our findings indicate that the roles of migfilin are functionally redundant during mouse development and tissue homeostasis.


Asunto(s)
Moléculas de Adhesión Celular/genética , Adhesión Celular/genética , Proteínas del Citoesqueleto/genética , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Integrina beta1/metabolismo , Animales , Moléculas de Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proteínas del Citoesqueleto/fisiología , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Proteínas con Dominio LIM , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...